全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

HIV-1 Reverse Transcriptase Still Remains a New Drug Target: Structure, Function, Classical Inhibitors, and New Inhibitors with Innovative Mechanisms of Actions

DOI: 10.1155/2012/586401

Full-Text   Cite this paper   Add to My Lib

Abstract:

During the retrotranscription process, characteristic of all retroviruses, the viral ssRNA genome is converted into integration-competent dsDNA. This process is accomplished by the virus-coded reverse transcriptase (RT) protein, which is a primary target in the current treatments for HIV-1 infection. In particular, in the approved therapeutic regimens two classes of drugs target RT, namely, nucleoside RT inhibitors (NRTIs) and nonnucleoside RT inhibitors (NNRTIs). Both classes inhibit the RT-associated polymerase activity: the NRTIs compete with the natural dNTP substrate and act as chain terminators, while the NNRTIs bind to an allosteric pocket and inhibit polymerization noncompetitively. In addition to these two classes, other RT inhibitors (RTIs) that target RT by distinct mechanisms have been identified and are currently under development. These include translocation-defective RTIs, delayed chain terminators RTIs, lethal mutagenesis RTIs, dinucleotide tetraphosphates, nucleotide-competing RTIs, pyrophosphate analogs, RT-associated RNase H function inhibitors, and dual activities inhibitors. This paper describes the HIV-1 RT function and molecular structure, illustrates the currently approved RTIs, and focuses on the mechanisms of action of the newer classes of RTIs. 1. Introduction Since the human immunodeficiency virus (HIV) has been established to be the etiological agent of the acquired immunodeficiency syndrome (AIDS) [1, 2], an originally unpredicted number of drugs have been approved for the treatment of the HIV-infected patients [3]. This success in effective drugs identification, certainly unique in the treatment of viral infections, together with the use of such armamentarium in different combination therapeutic regimens, has transformed a highly lethal syndrome into a chronic disease [4]. The management of this disease, however, is still complex and worrisome due to problems such as monitoring of therapy efficacy, chronic administration drug toxicity, poor tolerability, drug resistance development, or therapy adjustment after treatment failures [4]. For all these reasons, the search for new inhibitors, possibly acting with molecular mechanisms different from the ones of the already approved drugs or anyway showing different patterns of drug resistance and, possibly, with diverse drug-associated chronic toxicity, is still a worldwide health care issue. The success in HIV infection therapy is certainly related to the fact that the HIV life cycle has been intensely dissected; several of its steps have been validated as drug targets, and,

References

[1]  F. Barré-Sinoussi, J. C. Chermann, F. Rey et al., “Isolation of a T-lymphotropic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS),” Science, vol. 220, no. 4599, pp. 868–871, 1983.
[2]  S. Broder and R. C. Gallo, “A pathogenic retrovirus (HTLV-III) linked to AIDS,” New England Journal of Medicine, vol. 311, no. 20, pp. 1292–1297, 1984.
[3]  Y. Mehellou and E. De Clercq, “Twenty-six years of anti-HIV drug discovery: where do we stand and where do we go?” Journal of Medicinal Chemistry, vol. 53, no. 2, pp. 521–538, 2010.
[4]  A. M. N. Tsibris and M. S. Hirsch, “Antiretroviral therapy in the clinic,” Journal of Virology, vol. 84, no. 11, pp. 5458–5464, 2010.
[5]  L. Ratner, W. Haseltine, and R. Patarca, “Complete nucleotide sequence of the AIDS virus, HTLV-III,” Nature, vol. 313, no. 6000, pp. 277–284, 1985.
[6]  S. H. Hughes, E. Arnold, and Z. Hostomsky, “RNase H of retroviral reverse transcriptases,” in Ribonucleases H, R. J. Crouch and J. J. Toulmé, Eds., pp. 195–224, Les Editions Inserm, Paris, France, 1998.
[7]  H. E. Huber and C. C. Richardson, “Processing of the primer for plus strand DNA synthesis by human immunodeficiency virus 1 reverse transcriptase,” Journal of Biological Chemistry, vol. 265, no. 18, pp. 10565–10573, 1990.
[8]  J. W. Rausch and S. F. J. Le Grice, “'Binding, bending and bonding': polypurine tract-primed initiation of plus-strand DNA synthesis in human immunodeficiency virus,” International Journal of Biochemistry and Cell Biology, vol. 36, no. 9, pp. 1752–1766, 2004.
[9]  V. P. Basu, M. Song, L. Gao, S. T. Rigby, M. N. Hanson, and R. A. Bambara, “Strand transfer events during HIV-1 reverse transcription,” Virus Research, vol. 134, no. 1-2, pp. 19–38, 2008.
[10]  G. Divita, K. Rittinger, C. Geourjon, G. Deleage, and R. S. Goody, “Dimerization kinetics of HIV-1 and HIV-2 reverse transcriptase: a two step process,” Journal of Molecular Biology, vol. 245, no. 5, pp. 508–521, 1995.
[11]  L. A. Kohlstaedt, J. Wang, J. M. Friedman, P. A. Rice, and T. A. Steitz, “Crystal structure at 3.5 A resolution of HIV-1 reverse transcriptase complexes with an inhibitor,” Science, vol. 256, no. 5065, pp. 1783–1790, 1992.
[12]  A. Jacobo-Molina, J. Ding, R. G. Nanni et al., “Crystal structure of human immunodeficiency virus type 1 reverse transcriptase complexed with double-stranded DNA at 3.0 A resolution shows bent DNA,” Proceedings of the National Academy of Sciences of the United States of America, vol. 90, no. 13, pp. 6320–6324, 1993.
[13]  S. Liu, E. A. Abbondanzieri, J. W. Rausch, S. F. J. Le Grice, and X. Zhuang, “Slide into action: dynamic shuttling of HIV reverse transcriptase on nucleic acid substrates,” Science, vol. 322, no. 5904, pp. 1092–1097, 2008.
[14]  E. A. Abbondanzieri, G. Bokinsky, J. W. Rausch, J. X. Zhang, S. F. J. Le Grice, and X. Zhuang, “Dynamic binding orientations direct activity of HIV reverse transcriptase,” Nature, vol. 453, no. 7192, pp. 184–189, 2008.
[15]  T. A. Steltz, “A mechanism for all polymerases,” Nature, vol. 391, no. 6664, pp. 231–232, 1998.
[16]  M. Ghosh, P. S. Jacques, D. W. Rodgers, M. Ottman, J. L. Darlix, and S. F. J. Le Grice, “Alterations to the primer grip of p66 HIV-1 reverse transcriptase and their consequences for template-primer utilization,” Biochemistry, vol. 35, no. 26, pp. 8553–8562, 1996.
[17]  S. G. Sarafianos, B. Marchand, K. Das et al., “Structure and function of HIV-1 reverse transcriptase: molecular mechanisms of polymerization and inhibition,” Journal of Molecular Biology, vol. 385, no. 3, pp. 693–713, 2009.
[18]  M. W. Kellinger and K. A. Johnson, “Nucleotide-dependent conformational change governs specificity and analog discrimination by HIV reverse transcriptase,” Proceedings of the National Academy of Sciences of the United States of America, vol. 107, no. 17, pp. 7734–7739, 2010.
[19]  E. Tramontano and R. Di Santo, “HIV-1 RT-associated Rnase H function inhibitors: recent advances in drug development,” Current Medicinal Chemistry, vol. 17, no. 26, pp. 2837–2853, 2010.
[20]  M. Nowotny, S. A. Gaidamakov, R. J. Crouch, and W. Yang, “Crystal structures of RNase H bound to an RNA/DNA hybrid: substrate specificity and metal-dependent catalysis,” Cell, vol. 121, no. 7, pp. 1005–1016, 2005.
[21]  E. Rosta, M. Nowotny, W. Yang, and G. Hummer, “Catalytic mechanism of RNA backbone cleavage by ribonuclease H from quantum mechanics/molecular mechanics simulations,” Journal of the American Chemical Society, vol. 133, no. 23, pp. 8934–8941, 2011.
[22]  V. Mizrahi, M. T. Usdin, A. Harington, and L. R. Dudding, “Site-directed mutagenesis of the conserved Asp-443 and Asp-498 carboxy-terminal residues of HIV-1 reverse transcriptase,” Nucleic Acids Research, vol. 18, no. 18, pp. 5359–5363, 1990.
[23]  V. Mizrahi, R. L. Brooksbank, and N. C. Nkabinde, “Mutagenesis of the conserved aspartic acid 443, glutamic acid 478, asparagine 494, and aspartic acid 498 residues in the ribonuclease H domain of p66/p51 human immunodeficiency virus type I reverse transcriptase. Expression and biochemical analysis,” Journal of Biological Chemistry, vol. 269, no. 30, pp. 19245–19249, 1994.
[24]  G. L. Beilhartz and M. G?tte, “HIV-1 ribonuclease H: structure, catalytic mechanism and inhibitors,” Viruses, vol. 2, no. 4, pp. 900–926, 2010.
[25]  S. G. Sarafianos, K. Das, C. Tantillo et al., “Crystal structure of HIV-1 reverse transcriptase in complex with a polypurine tract RNA:DNA,” EMBO Journal, vol. 20, no. 6, pp. 1449–1461, 2001.
[26]  J. J. Champoux and S. J. Schultz, “Ribonuclease H: properties, substrate specificity and roles in retroviral reverse transcription,” FEBS Journal, vol. 276, no. 6, pp. 1506–1516, 2009.
[27]  E. S. Furfine and J. E. Reardon, “Reverse transcriptase. RNase H from the human immunodeficiency virus. Relationship of the DNA polymerase and RNA hydrolysis activities,” Journal of Biological Chemistry, vol. 266, no. 1, pp. 406–412, 1991.
[28]  A. Telesnitsky and S. P. Goff, “Reverse Transcriptase and the generation of retroviral DNA,” in Retroviruses, J. M. Coffin, S. H. Hughes, and H. E. Varmus, Eds., pp. 121–160, Cold Spring Harbor Laboratory Press, New York, NY, USA, 1997.
[29]  X. Ji, G. J. Klarmann, and B. D. Preston, “Effect of human immunodeficiency virus type 1 (HIV-1) nucleocapsid protein on HIV-1 reverse transcriptase activity in vitro,” Biochemistry, vol. 35, no. 1, pp. 132–143, 1996.
[30]  D. Grohmann, J. Godet, Y. Mély, J. L. Darlix, and T. Restle, “HIV-1 nucleocapsid traps reverse transcriptase on nucleic acid substrates,” Biochemistry, vol. 47, no. 46, pp. 12230–12240, 2008.
[31]  R. S. Aguiar and B. M. Peterlin, “APOBEC3 proteins and reverse transcription,” Virus Research, vol. 134, no. 1-2, pp. 74–85, 2008.
[32]  D. Arion, N. Kaushik, S. McCormick, G. Borkow, and M. A. Parniak, “Phenotypic mechanism of HIV-1 resistance to -azido- -deoxythymidine (AZT): increased polymerization processivity and enhanced sensitivity to pyrophosphate of the mutant viral reverse transcriptase,” Biochemistry, vol. 37, no. 45, pp. 15908–15917, 1998.
[33]  P. R. Meyer, S. E. Matsuura, A. G. So, and W. A. Scott, “Unblocking of chain-terminated primer by HIV-1 reverse transcriptase through a nucleotide-dependent mechanism,” Proceedings of the National Academy of Sciences of the United States of America, vol. 95, no. 23, pp. 13471–13476, 1998.
[34]  P. R. Meyer, S. E. Matsuura, A. Mohsin Mian, A. G. So, and W. A. Scott, “A mechanism of AZT resistance: an increase in nucleotide-dependent primer unblocking by mutant HIV-1 reverse transcriptase,” Molecular Cell, vol. 4, no. 1, pp. 35–43, 1999.
[35]  D. M. Simpson and M. Tagliati, “Nucleoside analogue-associated peripheral neuropathy in human immunodeficiency virus infection,” Journal of Acquired Immune Deficiency Syndromes and Human Retrovirology, vol. 9, no. 2, pp. 153–161, 1995.
[36]  R. F. Schinazi, R. M. Lloyd, M. H. Nguyen et al., “Characterization of human immunodeficiency viruses resistant to oxathiolane-cytosine nucleosides,” Antimicrobial Agents and Chemotherapy, vol. 37, no. 4, pp. 875–881, 1993.
[37]  R. Schuurman, M. Nijhuis, R. Van Leeuwen et al., “Rapid changes in human immunodeficiency virus type 1 RNA load and appearance of drug-resistant virus populations in persons treated with lamivudine (3TC),” Journal of Infectious Diseases, vol. 171, no. 6, pp. 1411–1419, 1995.
[38]  S. G. Sarafianos, K. Das, A. D. Clark et al., “Lamivudine (3TC) resistance in HIV-1 reverse transcriptase involves steric hindrance with β-branched amino acids,” Proceedings of the National Academy of Sciences of the United States of America, vol. 96, no. 18, pp. 10027–10032, 1999.
[39]  H. Huang, R. Chopra, G. L. Verdine, and S. C. Harrison, “Structure of a covalently trapped catalytic complex of HIV-1 reverse transcriptase: implications for drug resistance,” Science, vol. 282, no. 5394, pp. 1669–1675, 1998.
[40]  P. L. Boyer, S. G. Sarafianos, E. Arnold, and S. H. Hughes, “Selective excision of AZTMP by drug-resistant human immunodeficiency virus reverse transcriptase,” Journal of Virology, vol. 75, no. 10, pp. 4832–4842, 2001.
[41]  S. Dharmasena, Z. Pongracz, E. Arnold, S. G. Sarafianos, and M. A. Parniak, “ -azido- -deoxythymidine-( )-tetraphospho-( )-adenosine, the product of ATP-mediated excision of chain-terminating AZTMP, is a potent chain-terminating substrate for HIV-1 reverse transcriptase,” Biochemistry, vol. 46, no. 3, pp. 828–836, 2007.
[42]  N. Yahi, C. Tamalet, C. Tourrès, N. Tivoli, and J. Fantini, “Mutation L210W of HIV-1 reverse transcriptase in patients receiving combination therapy: incidence, association with other mutations, and effects on the structure of mutated reverse transcriptase,” Journal of Biomedical Science, vol. 7, no. 6, pp. 507–513, 2000.
[43]  G. N. Nikolenko, K. A. Delviks-Frankenberry, S. Palmer et al., “Mutations in the connection domain of HIV-1 reverse transcriptase increase -azido- -deoxythymidine resistance,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 1, pp. 317–322, 2007.
[44]  S. H. Yap, C. W. Sheen, J. Fahey et al., “N348I in the connection domain of HIV-1 reverse transcriptase confers zidovudine and nevirapine resistance,” PLoS Medicine, vol. 4, no. 12, Article ID e335, 2007.
[45]  K. A. Delviks-Frankenberry, G. N. Nikolenko, R. Barr, and V. K. Pathak, “Mutations in human immunodeficiency virus type 1 RNase H primer grip enhance -azido- -deoxythymidine resistance,” Journal of Virology, vol. 81, no. 13, pp. 6837–6845, 2007.
[46]  J. H. Brehm, D. Koontz, J. D. Meteer, V. Pathak, N. Sluis-Cremer, and J. W. Mellors, “Selection of mutations in the connection and RNase H domains of human immunodeficiency virus type 1 reverse transcriptase that increase resistance to -azido- -dideoxythymidine,” Journal of Virology, vol. 81, no. 15, pp. 7852–7859, 2007.
[47]  A. Hachiya, E. N. Kodama, S. G. Sarafianos et al., “Amino acid mutation N348I in the connection subdomain of human immunodeficiency virus type 1 reverse transcriptase confers multiclass resistance to nucleoside and nonnucleoside reverse transcriptase inhibitors,” Journal of Virology, vol. 82, no. 7, pp. 3261–3270, 2008.
[48]  S. Zelina, C. W. Sheen, J. Radzio, J. W. Mellors, and N. Sluis-Cremer, “Mechanisms by which the G333D mutation in human immunodeficiency virus type 1 reverse transcriptase facilitates dual resistance to zidovudine and lamivudine,” Antimicrobial Agents and Chemotherapy, vol. 52, no. 1, pp. 157–163, 2008.
[49]  K. A. Delviks-Frankenberry, G. N. Nikolenko, and V. K. Pathak, “The “connection” between HIV drug resistance and RNase H,” Viruses, vol. 2, no. 7, pp. 1476–1503, 2010.
[50]  J. Balzarini, L. Naesens, S. Aquaro et al., “Intracellular metabolism of CycloSaligenyl azido- , - dideoxythymidine monophosphate, a prodrug of -azido- , -dideoxythymidine (zidovudine),” Molecular Pharmacology, vol. 56, no. 6, pp. 1354–1361, 1999.
[51]  K. E. Squires, “An introduction to nucleoside and nucleotide analogues,” Antiviral Therapy, vol. 6, no. 3, pp. 1–14, 2001.
[52]  K. Das, S. E. Martinez, J. D. Bauman, and E. Arnold, “HIV-1 reverse transcriptase complex with DNA and nevirapine reveals non-nucleoside inhibition mechanism,” Nature Structural & Molecular Biology, vol. 19, pp. 253–259, 2012.
[53]  P. W. Mui, S. P. Jacober, K. D. Hargrave, and J. Adams, “Crystal structure of nevirapine, a non-nucleoside inhibitor of HIV-1 reverse transcriptase, and computational alignment with a structurally diverse inhibitor,” Journal of Medicinal Chemistry, vol. 35, no. 1, pp. 201–202, 1992.
[54]  W. Sch?fer, W. G. Friebe, H. Leinert et al., “Non-nucleoside inhibitors of HIV-1 reverse transcriptase: molecular modeling and X-ray structure investigations,” Journal of Medicinal Chemistry, vol. 36, no. 6, pp. 726–732, 1993.
[55]  J. Ding, K. Das, C. Tantillo et al., “Structure of HIV-1 reverse transcriptase in a complex with the non-nucleoside inhibitor α-APA R 95845 at 2.8 A resolution,” Structure, vol. 3, no. 4, pp. 365–379, 1995.
[56]  P. P. Mager, “Hybrid canonical-correlation neural-network approach applied to nonnucleoside HIV-1 reverse transcriptase inhibitors (HEPT derivatives),” Current Medicinal Chemistry, vol. 10, no. 17, pp. 1643–1659, 2003.
[57]  N. Sluis-Cremer, N. A. Temiz, and I. Bahar, “Conformational changes in HIV-1 reverse transcriptase induced by nonnucleoside reverse transcriptase inhibitor binding,” Current HIV Research, vol. 2, no. 4, pp. 323–332, 2004.
[58]  E. De Clercq, “Perspectives of non-nucleoside reverse transcriptase inhibitors (NNRTIs) in the therapy of HIV-1 infection,” Farmaco, vol. 54, no. 1-2, pp. 26–45, 1999.
[59]  J. Balzarini, “Current status of the non-nucleoside reverse transcriptase inhibitors of human immunodeficiency virus type 1,” Current Topics in Medicinal Chemistry, vol. 4, no. 9, pp. 921–944, 2004.
[60]  R. M. Esnouf, J. Ren, A. L. Hopkins et al., “Unique features in the structure of the complex between HIV-1 reverse transcriptase and the bis(heteroaryl)piperazine (BHAP) U-90152 explain resistance mutations for this nonnucleoside inhibitor,” Proceedings of the National Academy of Sciences of the United States of America, vol. 94, no. 8, pp. 3984–3989, 1997.
[61]  J. Ren and D. K. Stammers, “Structural basis for drug resistance mechanisms for non-nucleoside inhibitors of HIV reverse transcriptase,” Virus Research, vol. 134, no. 1-2, pp. 157–170, 2008.
[62]  J. W. Mellors, G. E. Dutschman, G. J. Im, E. Tramontano, S. R. Winkler, and Y. C. Cheng, “In vitro selection and molecular characterization of human immunodeficiency virus-1 resistant to non-nucleoside inhibitors of reverse transcriptase,” Molecular Pharmacology, vol. 41, no. 3, pp. 446–451, 1992.
[63]  J. W. Mellors, G. J. Im, E. Tramontano et al., “A single conservative amino acid substitution in the reverse transcriptase of human immunodeficiency virus-1 confers resistance to (+)-(5S)-4,5,6,7- tetrahydro-5-methyl-6-(3-methyl-2-butenyl)imidazo[4,5,1- jk][1,4]benzodiazepin-2(1H)-thione (TIBO R82150),” Molecular Pharmacology, vol. 43, no. 1, pp. 11–16, 1993.
[64]  G. N. Nikolenko, K. A. Delviks-Frankenberry, and V. K. Pathak, “A novel molecular mechanism of dual resistance to nucleoside and nonnucleoside reverse transcriptase inhibitors,” Journal of Virology, vol. 84, no. 10, pp. 5238–5249, 2010.
[65]  T. Cihlar and A. S. Ray, “Nucleoside and nucleotide HIV reverse transcriptase inhibitors: 25 years after zidovudine,” Antiviral Research, vol. 85, no. 1, pp. 39–58, 2010.
[66]  H. T. Ho and M. J. Hitchcock, “Cellular pharmacology of , -dideoxy- , -didehydrothymidine, a nucleoside analog active against human immunodeficiency virus,” Antimicrobial Agents and Chemotherapy, vol. 33, no. 6, pp. 844–849, 1989.
[67]  L. J. Waters, G. Moyle, S. Bonora et al., “Abacavir plasma pharmacokinetics in the absence and presence of atazanavir/ritonavir or lopinavir/ritonavir and vice versa in HIV-infected patients,” Antiviral Therapy, vol. 12, no. 5, pp. 825–830, 2007.
[68]  J. A. Mcdowell, G. E. Chittick, C. P. Stevens, K. D. Edwards, and D. S. Stein, “Pharmacokinetic interaction of abacavir (1592U89) and ethanol in human immunodeficiency virus-infected adults,” Antimicrobial Agents and Chemotherapy, vol. 44, no. 6, pp. 1686–1690, 2000.
[69]  J. A. Johnson, J. F. Li, X. Wei et al., “Minority HIV-1 drug resistance mutations are present in antiretroviral treatment-naive populations and associate with reduced treatment efficacy,” PLoS Medicine, vol. 5, no. 7, Article ID e158, 2008.
[70]  R. C. Bethell, Y. S. Lie, and N. T. Parkin, “In vitro activity of SPD754, a new deoxycytidine nucleoside reverse transcriptase inhibitor (NRTI), against 215 HIV-1 isolates resistant to other NRTIs,” Antiviral Chemistry and Chemotherapy, vol. 16, no. 5, pp. 295–302, 2005.
[71]  Z. Gu, B. Allard, J. M. De Muys et al., “In vitro antiretroviral activity and in vitro toxicity profile of SPD754, a new deoxycytidine nucleoside reverse transcriptase inhibitor for treatment of human immunodeficiency virus infection,” Antimicrobial Agents and Chemotherapy, vol. 50, no. 2, pp. 625–631, 2006.
[72]  S. Cox and J. Southby, “Apricitabine—a novel nucleoside reverse transcriptase inhibitor for the treatment of HIV infection that is refractory to existing drugs,” Expert Opinion on Investigational Drugs, vol. 18, no. 2, pp. 199–209, 2009.
[73]  M. P. de Baar, E. R. de Rooij, K. G. M. Smolders, H. B. van Schijndel, E. C. Timmermans, and R. Bethell, “Effects of apricitabine and other nucleoside reverse transcriptase inhibitors on replication of mitochondrial DNA in HepG2 cells,” Antiviral Research, vol. 76, no. 1, pp. 68–74, 2007.
[74]  R. Bethell, J. De Muys, J. Lippens et al., “In vitro interactions between apricitabine and other deoxycytidine analogues,” Antimicrobial Agents and Chemotherapy, vol. 51, no. 8, pp. 2948–2953, 2007.
[75]  T. S. Lin, M. Z. Luo, M. C. Liu et al., “Design and synthesis of , -dideoxy- , -didehydro-β-L-cytidine (β- L-d4C) and ,3'-dideoxy- - -didehydro-β-L-5-fluorocytidine (β-L-Fd4C), two exceptionally potent inhibitors of human hepatitis B virus (HBV) and potent inhibitors of human immunodeficiency virus (HIV) in vitro,” Journal of Medicinal Chemistry, vol. 39, no. 9, pp. 1757–1759, 1996.
[76]  G. E. Dutschman, E. G. Bridges, S. H. Liu et al., “Metabolism of , -dideoxy- , -didehydro-β-L(-)-5-fluorocytidine and its activity in combination with clinically approved anti-human immunodeficiency virus β-D(+) nucleoside analogs in vitro,” Antimicrobial Agents and Chemotherapy, vol. 42, no. 7, pp. 1799–1804, 1998.
[77]  J. L. Hammond, U. M. Parikh, D. L. Koontz et al., “In vitro selection and analysis of human immunodeficiency virus type 1 resistant to derivatives of β- , -didehydro- , -dideoxy-5-fluorocytidine,” Antimicrobial Agents and Chemotherapy, vol. 49, no. 9, pp. 3930–3932, 2005.
[78]  U. M. Parikh, D. L. Koontz, C. K. Chu, R. F. Schinazi, and J. W. Mellors, “In vitro activity of structurally diverse nucleoside analogs against human immunodeficiency virus type 1 with the K65R mutation in reverse transcriptase,” Antimicrobial Agents and Chemotherapy, vol. 49, no. 3, pp. 1139–1144, 2005.
[79]  Z. Gu, M. A. Wainberg, N. Nguyen-Ba et al., “Mechanism of action and in vitro activity of , -dioxolanylpurine nucleoside analogues against sensitive and drug-resistant human immunodeficiency virus type 1 variants,” Antimicrobial Agents and Chemotherapy, vol. 43, no. 10, pp. 2376–2382, 1999.
[80]  J. P. Mewshaw, F. T. Myrick, D. A. C. S. Wakefield et al., “Dioxolane guanosine, the active form of the prodrug diaminopurine dioxolane, is a potent inhibitor of drug-resistant HIV-1 isolates from patients for whom standard nucleoside therapy fails,” Journal of Acquired Immune Deficiency Syndromes, vol. 29, no. 1, pp. 11–20, 2002.
[81]  H. Z. Bazmi, J. L. Hammond, S. C. H. Cavalcanti, C. K. Chu, R. F. Schinazi, and J. W. Mellors, “In vitro selection of mutations in the human immunodeficiency virus type 1 reverse transcriptase that decrease susceptibility to (-)-β-D-dioxolane- guanosine and suppress resistance to -azido- -deoxythymidine,” Antimicrobial Agents and Chemotherapy, vol. 44, no. 7, pp. 1783–1788, 2000.
[82]  P. A. Furman, J. Jeffrey, L. L. Kiefer et al., “Mechanism of action of 1-β-D-2,6-diaminopurine dioxolane, a prodrug of the human immunodeficiency virus type 1 inhibitor 1-β-D-dioxolane guanosine,” Antimicrobial Agents and Chemotherapy, vol. 45, no. 1, pp. 158–165, 2001.
[83]  J. Y. Feng and K. S. Anderson, “Mechanistic studies comparing the incorporation of (+) and (-) isomers of 3TCTP by HIV-1 reverse transcriptase,” Biochemistry, vol. 38, no. 1, pp. 55–63, 1999.
[84]  D. S. Shewach, D. C. Liotta, and R. F. Schinazi, “Affinity of the antiviral enantiomers of oxathiolane cytosine nucleosides for human -deoxycytidine kinase,” Biochemical Pharmacology, vol. 45, no. 7, pp. 1540–1543, 1993.
[85]  R. F. Schinazi, A. McMillan, D. Cannon et al., “Selective inhibition of human immunodeficiency viruses by racemates and enantiomers of cis-5-fluoro-1-[2-(hydroxymethyl)-1,3-oxathiolan-5-yl]cytosine,” Antimicrobial Agents and Chemotherapy, vol. 36, no. 11, pp. 2423–2431, 1992.
[86]  R. F. Schinazi, A. McMillan, R. L. Lloyd, S. Schlueter-Wirtz, D. C. Liotta, and C. K. Chu, “Molecular properties of HIV-1 resistant to (+)-enantiomers and racemates of oxathiolane cytosine nucleosides and their potential for the treatment of HIV and HBV infections,” Antiviral Research, vol. 34, article A42, 1997.
[87]  C. Herzmann, K. Arastèh, R. L. Murphy et al., “Safety, pharmacokinetics, and efficacy of (+/-)-β- , - dideoxy-5-fluoro- -thiacytidine with efavirenz and stavudine in antiretroviral-naive human immunodeficiency virus-infected patients,” Antimicrobial Agents and Chemotherapy, vol. 49, no. 7, pp. 2828–2833, 2005.
[88]  J. Y. Feng, E. Murakami, S. M. Zorca et al., “Relationship between antiviral activity and host toxicity: comparison of the incorporation efficiencies of , -Dideoxy-5-Fluoro- -Thiacytidine-triphosphate analogs by human immunodeficiency virus type 1 reverse transcriptase and human mitochondrial DNA polymerase,” Antimicrobial Agents and Chemotherapy, vol. 48, no. 4, pp. 1300–1306, 2004.
[89]  G. E. Dutschman, S. P. Grill, E. A. Gullen et al., “Novel -substituted stavudine analog with improved anti-human immunodeficiency virus activity and decreased cytotoxicity,” Antimicrobial Agents and Chemotherapy, vol. 48, no. 5, pp. 1640–1646, 2004.
[90]  P. Herdewijn, J. Balzarini, M. Baba et al., “Synthesis and anti-HIV activity of different sugar-modified pyrimidine and purine nucleosides,” Journal of Medicinal Chemistry, vol. 31, no. 10, pp. 2040–2048, 1988.
[91]  H. Hayakawa, S. Kohgo, K. Kitano et al., “Potential of -C-substituted nucleosides for the treatment of HIV-1,” Antiviral Chemistry and Chemotherapy, vol. 15, no. 4, pp. 169–187, 2004.
[92]  A. Kawamoto, E. Kodama, S. G. Sarafianos et al., “ -Deoxy- -C-ethynyl-2-halo-adenosines active against drug-resistant human immunodeficiency virus type 1 variants,” International Journal of Biochemistry and Cell Biology, vol. 40, no. 11, pp. 2410–2420, 2008.
[93]  E. Michailidis, B. Marchand, E. N. Kodama et al., “Mechanism of inhibition of HIV-1 reverse transcriptase by -ethynyl-2-fluoro- -deoxyadenosine triphosphate, a translocation-defective reverse transcriptase inhibitor,” Journal of Biological Chemistry, vol. 284, no. 51, pp. 35681–35691, 2009.
[94]  K. A. Kirby, K. Singh, E. Michailidis et al., “The sugar ring conformation of -ethynyl-2-fluoro- -deoxyadenosine and its recognition by the polymerase active site of HIV reverse transcriptase,” Cellular and Molecular Biology, vol. 57, no. 1, pp. 40–46, 2011.
[95]  C. D. Sohl, K. Singh, R. Kasiviswanathan et al., “The mechanism of interaction of human mitochondrial DNA γ with the novel nucleoside reverse transcriptase inhibitor -Ethynyl-2-Fluoro- -deoxyadenosine indicates a low potential for host toxicity,” Antimicrobial Agents and Chemotherapy, vol. 56, pp. 1630–1634, 2012.
[96]  V. Vivet?Boudou, C. Isel, M. Sleiman et al., “8-modified- -deoxyadenosine analogues induce delayed polymerization arrest during HIV-1 reverse transcription,” PLoS ONE, vol. 6, no. 11, Article ID e27456, 2011.
[97]  L. A. Loeb, J. M. Essigmann, F. Kazazi, J. Zhang, K. D. Rose, and J. I. Mullins, “Lethal mutagenesis of HIV with mutagenic nucleoside analogs,” Proceedings of the National Academy of Sciences of the United States of America, vol. 96, no. 4, pp. 1492–1497, 1999.
[98]  K. S. Harris, W. Brabant, S. Styrchak, A. Gall, and R. Daifuku, “KP-1212/1461, a nucleoside designed for the treatment of HIV by viral mutagenesis,” Antiviral Research, vol. 67, no. 1, pp. 1–9, 2005.
[99]  R. A. Smith, L. A. Loeb, and B. D. Preston, “Lethal mutagenesis of HIV,” Virus Research, vol. 107, no. 2, pp. 215–228, 2005.
[100]  X. Tu, K. Das, Q. Han et al., “Structural basis of HIV-1 resistance to AZT by excision,” Nature Structural and Molecular Biology, vol. 17, no. 10, pp. 1202–1209, 2010.
[101]  P. R. Meyer, A. J. Smith, S. E. Matsuura, and W. A. Scott, “Chain-terminating dinucleoside tetraphosphates are substrates for DNA polymerization by human immunodeficiency virus type 1 reverse transcriptase with increased activity against thymidine analogue-resistant mutants,” Antimicrobial Agents and Chemotherapy, vol. 50, no. 11, pp. 3607–3614, 2006.
[102]  N. Sluis-Cremer and G. Tachedjian, “Mechanisms of inhibition of HIV replication by non-nucleoside reverse transcriptase inhibitors,” Virus Research, vol. 134, no. 1-2, pp. 147–156, 2008.
[103]  P. Zhan, X. Chen, D. Li, Z. Fang, E. De Clercq, and X. Liu, “HIV-1 NNRTIs: structural diversity, pharmacophore similarity, and implications for drug design,” Medicinal Research Reviews. In press.
[104]  K. Das, P. J. Lewi, S. H. Hughes, and E. Arnold, “Crystallography and the design of anti-AIDS drugs: conformational flexibility and positional adaptability are important in the design of non-nucleoside HIV-1 reverse transcriptase inhibitors,” Progress in Biophysics and Molecular Biology, vol. 88, no. 2, pp. 209–231, 2005.
[105]  P. Zhan, X. Liu, Z. Li, C. Pannecouque, and E. De Clercq, “Design strategies of novel NNRTIs to overcome drug resistance,” Current Medicinal Chemistry, vol. 16, no. 29, pp. 3903–3917, 2009.
[106]  P. A. J. Janssen, P. J. Lewi, E. Arnold et al., “In search of a novel anti-HIV drug: multidisciplinary coordination in the discovery of 4-[[4-[[4-[(1E)-2-cyanoethenyl]-2,6-dimethylphenyl]amino]-2- pyrimidinyl]amino]benzonitrile (R278474, rilpivirine),” Journal of Medicinal Chemistry, vol. 48, no. 6, pp. 1901–1909, 2005.
[107]  J. D. Pata, W. G. Stirtan, S. W. Goldstein, and T. A. Steitz, “Structure of HIV-1 reverse transcriptase bound to an inhibitor active against mutant reverse transcriptases resistant to other nonnucleoside inhibitors,” Proceedings of the National Academy of Sciences of the United States of America, vol. 101, no. 29, pp. 10548–10553, 2004.
[108]  J. Ren and D. K. Stammers, “HIV reverse transcriptase structures: designing new inhibitors and understanding mechanisms of drug resistance,” Trends in Pharmacological Sciences, vol. 26, no. 1, pp. 4–7, 2005.
[109]  J. Tang, K. Maddali, C. D. Dreis et al., “N-3 hydroxylation of pyrimidine-2,4-diones yields dual inhibitors of HIV reverse transcriptase and integrase,” ACS Medicinal Chemistry Letters, vol. 2, no. 1, pp. 63–67, 2011.
[110]  J. Tang, K. Maddali, C. D. Dreis et al., “6-Benzoyl-3-hydroxypyrimidine-2,4-diones as dual inhibitors of HIV reverse transcriptase and integrase,” Bioorganic and Medicinal Chemistry Letters, vol. 21, no. 8, pp. 2400–2402, 2011.
[111]  D. Jochmans, J. Deval, B. Kesteleyn et al., “Indolopyridones inhibit human immunodeficiency virus reverse transcriptase with a novel mechanism of action,” Journal of Virology, vol. 80, no. 24, pp. 12283–12292, 2006.
[112]  Z. Zhang, M. Walker, W. Xu et al., “Novel nonnucleoside inhibitors that select nucleoside inhibitor resistance mutations in human immunodeficiency virus type 1 reverse transcriptase,” Antimicrobial Agents and Chemotherapy, vol. 50, no. 8, pp. 2772–2781, 2006.
[113]  M. Ehteshami, B. J. Scarth, E. P. Tchesnokov et al., “Mutations M184V and Y115F in HIV-1 reverse transcriptase discriminate against “nucleotide-competing reverse transcriptase inhibitors”,” Journal of Biological Chemistry, vol. 283, no. 44, pp. 29904–29911, 2008.
[114]  A. Auger, G. L. Beilhartz, S. Zhu et al., “Impact of primer-induced conformational dynamics of HIV-1 reverse transcriptase on polymerase translocation and inhibition,” The Journal of Biological Chemistry, vol. 286, pp. 29575–29583, 2011.
[115]  G. Maga, M. Radi, S. Zanoli et al., “Discovery of non-nucleoside inhibitors of HIV-1 reverse transcriptase competing with the nucleotide substrate,” Angewandte Chemie, vol. 46, no. 11, pp. 1810–1813, 2007.
[116]  M. Radi, C. Falciani, L. Contemori et al., “A multidisciplinary approach for the identification of novel HIV-1 non-nucleoside reverse transcriptase inhibitors: S-DABOCs and DAVPs,” ChemMedChem, vol. 3, no. 4, pp. 573–593, 2008.
[117]  S. Freisz, G. Bec, M. Radi et al., “Crystal structure of HIV-1 reverse transcriptase bound to a non-nucleoside inhibitor with a novel mechanism of action,” Angewandte Chemie, vol. 49, no. 10, pp. 1805–1808, 2010.
[118]  B. Oberg, “Antiviral effects of phosphonoformate (PFA, foscarnet sodium),” Pharmacology and Therapeutics, vol. 40, no. 2, pp. 213–285, 1989.
[119]  R. R. Razonable, “Antiviral drugs for viruses other than human immunodeficiency virus,” Mayo Clinic Proceedings, vol. 86, pp. 1009–1026, 2011.
[120]  D. Derse, K. F. Bastow, and Y. Cheng, “Characterization of the DNA polymerases induced by a group of herpes simplex virus type I variants selected for growth in the presence of phosphonoformic acid,” Journal of Biological Chemistry, vol. 257, no. 17, pp. 10251–10260, 1982.
[121]  B. Marchand, E. P. Tchesnokov, and M. G?tte, “The pyrophosphate analogue foscarnet traps the pre-translocational state of HIV-1 reverse transcriptase in a Brownian ratchet model of polymerase translocation,” Journal of Biological Chemistry, vol. 282, no. 5, pp. 3337–3346, 2007.
[122]  J. W. Mellors, H. Z. Bazmi, R. F. Schinazi et al., “Novel mutations in reverse transcriptase of human immunodeficiency virus type 1 reduce susceptibility to foscarnet in laboratory and clinical isolates,” Antimicrobial Agents and Chemotherapy, vol. 39, no. 5, pp. 1087–1092, 1995.
[123]  G. Tachedjian, D. J. Hooker, A. D. Gurusinghe et al., “Characterisation of foscarnet-resistant strains of human immunodeficiency virus type 1,” Virology, vol. 212, no. 1, pp. 58–68, 1995.
[124]  G. J. Im, E. Tramontano, C. J. Gonzalez, and Y. C. Cheng, “Identification of the amino acid in the human immunodeficiency virus type 1 reverse transcriptase involved in the pyrophosphate binding of antiviral nucleoside triphosphate analogs and phosphonoformate. Implications for multiple drug resistance,” Biochemical Pharmacology, vol. 46, no. 12, pp. 2307–2313, 1993.
[125]  E. Tramontano, G. Piras, J. W. Mellors, M. Putzolu, H. Z. Bazmi, and P. La Colla, “Biochemical characterization of HIV-1 reverse transcriptases encoding mutations at amino acid residues 161 and 208 involved in resistance to phosphonoformate,” Biochemical Pharmacology, vol. 56, no. 12, pp. 1583–1589, 1998.
[126]  C. Cruchaga, E. Ansó, A. Rouzaut, and J. J. Martínez-Irujo, “Selective excision of chain-terminating nucleotides by HIV-1 reverse transcriptase with phosphonoformate as substrate,” Journal of Biological Chemistry, vol. 281, no. 38, pp. 27744–27752, 2006.
[127]  E. Tramontano, “HIV-1 RNase H: recent progress in an exciting, yet little explored, drug target,” Mini-Reviews in Medicinal Chemistry, vol. 6, no. 6, pp. 727–737, 2006.
[128]  E. B. Lansdon, Q. Liu, S. A. Leavitt et al., “Structural and binding analysis of pyrimidinol carboxylic acid and N-hydroxy quinazolinedione HIV-1 RNase H inhibitors,” Antimicrobial Agents and Chemotherapy, vol. 55, no. 6, pp. 2905–2915, 2011.
[129]  H. Fuji, E. Urano, Y. Futahashi et al., “Derivatives of 5-nitro-furan-2-carboxylic acid carbamoylmethyl ester inhibit RNase H activity associated with HIV-1 reverse transcriptase,” Journal of Medicinal Chemistry, vol. 52, no. 5, pp. 1380–1387, 2009.
[130]  H. Yanagita, E. Urano, K. Matsumoto et al., “Structural and biochemical study on the inhibitory activity of derivatives of 5-nitro-furan-2-carboxylic acid for RNase H function of HIV-1 reverse transcriptase,” Bioorganic and Medicinal Chemistry, vol. 19, no. 2, pp. 816–825, 2011.
[131]  H. P. Su, Y. Yan, G. S. Prasad et al., “Structural basis for the inhibition of RNase H activity of HIV-1 reverse transcriptase by RNase H active site-directed inhibitors,” Journal of Virology, vol. 84, no. 15, pp. 7625–7633, 2010.
[132]  C. A. Shaw-Reid, V. Munshi, P. Graham et al., “Inhibition of HIV-1 ribonuclease H by a novel diketo acid, 4-[5-(benzoylamino)thien-2-yl]-2,4-dioxobutanoic acid,” Journal of Biological Chemistry, vol. 278, no. 5, pp. 2777–2780, 2003.
[133]  E. Tramontano, F. Esposito, R. Badas, R. Di Santo, R. Costi, and P. La Colla, “6-[1-(4-Fluorophenyl)methyl-1H-pyrrol-2-yl)]-2,4-dioxo-5-hexenoic acid ethyl ester a novel diketo acid derivative which selectively inhibits the HIV-1 viral replication in cell culture and the ribonuclease H activity in vitro,” Antiviral Research, vol. 65, no. 2, pp. 117–124, 2005.
[134]  R. Di Santo, R. Costi, M. Artico, E. Tramontano, P. La Colla, and A. Pani, “HIV-1 integrase inhibitors that block HIV-1 replication in infected cells. Planning synthetic derivatives from natural products,” Pure and Applied Chemistry, vol. 75, no. 2-3, pp. 195–206, 2003.
[135]  R. Costi, R. Di Santo, M. Artico et al., “6-Aryl-2,4-dioxo-5-hexenoic acids, novel integrase inhibitors active against HIV-1 multiplication in cell-based assays,” Bioorganic and Medicinal Chemistry Letters, vol. 14, no. 7, pp. 1745–1749, 2004.
[136]  R. Costi, R. Di Santo, M. Artico et al., “2,6-Bis(3,4,5-trihydroxybenzylydene) derivatives of cyclohexanone: novel potent HIV-1 integrase inhibitors that prevent HIV-1 multiplication in cell-based assays,” Bioorganic and Medicinal Chemistry, vol. 12, no. 1, pp. 199–215, 2004.
[137]  K. Klumpp, J. Q. Hang, S. Rajendran et al., “Two-metal ion mechanism of RNA cleavage by HIV RNase H and mechanism-based design of selective HIV RNase H inhibitors,” Nucleic Acids Research, vol. 31, no. 23, pp. 6852–6859, 2003.
[138]  J. Q. Hang, S. Rajendran, Y. Yang et al., “Activity of the isolated HIV RNase H domain and specific inhibition by N-hydroxyimides,” Biochemical and Biophysical Research Communications, vol. 317, no. 2, pp. 321–329, 2004.
[139]  M. Billamboz, F. Bailly, M. L. Barreca et al., “Design, synthesis, and biological evaluation of a series of 2-hydroxyisoquinoline-1,3(2H,4H)-diones as dual inhibitors of human immunodeficiency virus type 1 integrase and the reverse transcriptase RNase H domain,” Journal of Medicinal Chemistry, vol. 51, no. 24, pp. 7717–7730, 2008.
[140]  M. Billamboz, F. Bailly, C. Lion et al., “Magnesium chelating 2-hydroxyisoquinoline-1,3(2H, 4H)-diones, as inhibitors of HIV-1 integrase and/or the HIV-1 reverse transcriptase ribonuclease H domain: discovery of a novel selective inhibitor of the ribonuclease H function,” Journal of Medicinal Chemistry, vol. 54, no. 6, pp. 1812–1824, 2011.
[141]  M. Wendeler, H. F. Lee, A. Bermingham et al., “Vinylogous ureas as a novel class of inhibitors of reverse transcriptase-associated ribonuclease H activity,” ACS Chemical Biology, vol. 3, no. 10, pp. 635–644, 2008.
[142]  S. Chung, M. Wendeler, J. W. Rausch et al., “Structure-activity analysis of vinylogous urea inhibitors of human immunodeficiency virus-encoded ribonuclease H,” Antimicrobial Agents and Chemotherapy, vol. 54, no. 9, pp. 3913–3921, 2010.
[143]  G. Borkow, R. S. Fletcher, J. Barnard et al., “Inhibition of the ribonuclease H and DNA polymerase activities of HIV- 1 reverse transcriptase by N-(4-tert-butylbenzoyl)-2-hydroxy-1- naphthaldehyde hydrazone,” Biochemistry, vol. 36, no. 11, pp. 3179–3185, 1997.
[144]  D. Arion, N. Sluis-Cremer, K. L. Min, M. E. Abram, R. S. Fletcher, and M. A. Parniak, “Mutational analysis of tyr-501 of HIV-1 reverse transcriptase: effects on ribonuclease H activity and inhibition of this activity by N-acylhydrazones,” Journal of Biological Chemistry, vol. 277, no. 2, pp. 1370–1374, 2002.
[145]  N. Sluis-Cremer, D. Arion, and M. A. Parniak, “Destabilization of the HIV-1 reverse transcriptase dimer upon interaction with N-acyl hydrazone inhibitors,” Molecular Pharmacology, vol. 62, no. 2, pp. 398–405, 2002.
[146]  D. M. Himmel, S. G. Sarafianos, S. Dharmasena et al., “HIV-1 reverse transcriptase structure with RNase H inhibitor dihydroxy benzoyl naphthyl hydrazone bound at a novel site,” ACS Chemical Biology, vol. 1, no. 11, pp. 702–712, 2006.
[147]  A. K. Felts, K. La Barge, J. D. Bauman et al., “Identification of alternative binding sites for inhibitors of HIV-1 ribonuclease H through comparative analysis of virtual enrichment studies,” Journal of Chemical Information and Modeling, vol. 51, no. 5, pp. 1986–1998, 2011.
[148]  K. Tatyana, E. Francesca, Z. Luca et al., “Inhibition of HIV-1 ribonuclease H activity by novel frangula-emodine derivatives,” Medicinal Chemistry, vol. 5, no. 5, pp. 398–410, 2009.
[149]  F. Esposito, T. Kharlamova, S. Distinto et al., “Alizarine derivatives as new dual inhibitors of the HIV-1 reverse transcriptase-associated DNA polymerase and RNase H activities effective also on the RNase H activity of non-nucleoside resistant reverse transcriptases,” FEBS Journal, vol. 278, no. 9, pp. 1444–1457, 2011.
[150]  E. Tramontano, T. Kharlamova, and F. Esposito, “Effect of new quinizarin derivatives on both HCV NS5B RNA polymerase and HIV-1 reverse transcriptase associated ribonuclease H activities,” Journal of Chemotherapy, vol. 23, pp. 273–276, 2011.
[151]  P. Mohan, S. Loya, O. Avidan et al., “Synthesis of naphthalenesulfonic acid small molecules as selective inhibitors of the DNA polymerase and ribonuclease H activities of HIV-1 reverse transcriptase,” Journal of Medicinal Chemistry, vol. 37, no. 16, pp. 2513–2519, 1994.
[152]  A. G. Skillman, K. W. Maurer, D. C. Roe et al., “A novel mechanism for inhibition of HIV-1 reverse transcriptase,” Bioorganic Chemistry, vol. 30, no. 6, pp. 443–458, 2002.
[153]  L. Z. Wang, G. L. Kenyon, and K. A. Johnson, “Novel mechanism of inhibition of HIV-1 reverse transcriptase by a new non-nucleoside analog, KM-1,” Journal of Biological Chemistry, vol. 279, no. 37, pp. 38424–38432, 2004.
[154]  C. A. Davis, M. A. Parniak, and S. H. Hughes, “The effects of RNase H inhibitors and nevirapine on the susceptibility of HIV-1 to AZT and 3TC,” Virology, vol. 419, pp. 64–71, 2011.
[155]  S. Srivastava, N. Sluis-Cremer, and G. Tachedjian, “Dimerization of human immunodeficiency virus type 1 reverse transcriptase as an antiviral target,” Current Pharmaceutical Design, vol. 12, no. 15, pp. 1879–1894, 2006.
[156]  M. J. Camarasa, S. Velázquez, A. San-Félix, M. J. Pérez-Pérez, and F. Gago, “Dimerization inhibitors of HIV-1 reverse transcriptase, protease and integrase: a single mode of inhibition for the three HIV enzymes?” Antiviral Research, vol. 71, no. 2-3, pp. 260–267, 2006.
[157]  D. Grohmann, V. Corradi, M. Elbasyouny et al., “Small molecule inhibitors targeting HIV-1 reverse transcriptase dimerization,” ChemBioChem, vol. 9, no. 6, pp. 916–922, 2008.
[158]  K. Warren, D. Warrilow, L. Meredith, and D. Harrich, “Reverse transcriptase and cellular factors: regulators of HIV-1 reverse transcriptase,” Viruses, vol. 1, pp. 873–894, 2009.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413